Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/322051216

The blastema and epimorphic regeneration in mammals

Article  in  Developmental Biology · December 2017


DOI: 10.1016/j.ydbio.2017.08.007

CITATIONS READS

20 702

2 authors:

Ashley Seifert Ken Muneoka


University of Kentucky Texas A&M University
59 PUBLICATIONS   1,092 CITATIONS    103 PUBLICATIONS   3,370 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Mammalian Regeneration View project

Spiny mouse sleep analysis View project

All content following this page was uploaded by Ken Muneoka on 07 February 2018.

The user has requested enhancement of the downloaded file.


Developmental Biology 433 (2018) 190–199

Contents lists available at ScienceDirect

Developmental Biology
journal homepage: www.elsevier.com/locate/developmentalbiology

Review article

The blastema and epimorphic regeneration in mammals MARK


a,⁎ b,⁎
Ashley W. Seifert , Ken Muneoka
a
Department of Biology, University of Kentucky, Lexington, KY 40506, United States
b
Department of Veterinary Physiology and Pharmacology, Texas A & M University, College Station, TX 77843, United States

A BS T RAC T

Studying regeneration in animals where and when it occurs is inherently interesting and a challenging research
topic within developmental biology. Historically, vertebrate regeneration has been investigated in animals that
display enhanced regenerative abilities and we have learned much from studying organ regeneration in
amphibians and fish. From an applied perspective, while regeneration biologists will undoubtedly continue to
study poikilothermic animals (i.e., amphibians and fish), studies focused on homeotherms (i.e., mammals and
birds) are also necessary to advance regeneration biology. Emerging mammalian models of epimorphic
regeneration are poised to help link regenerative biology and regenerative medicine. The regenerating rodent
digit tip, which parallels human fingertip regeneration, and the regeneration of large circular defects through
the ear pinna in spiny mice and rabbits, provide tractable, experimental systems where complex tissue
structures are regrown through blastema formation and morphogenesis. Using these models as examples, we
detail similarities and differences between the mammalian blastema and its classical counterpart to arrive at a
broad working definition of a vertebrate regeneration blastema. This comparison leads us to conclude that
regenerative failure is not related to the availability of regeneration-competent progenitor cells, but is most
likely a function of the cellular response to the microenvironment that forms following traumatic injury. Recent
studies demonstrating that targeted modification of this microenvironment can restrict or enhance regenerative
capabilities in mammals helps provide a roadmap for eventually pushing the limits of human regeneration.

1. Introduction clinical problem, although all tissues of the human body display some
level of homeostatic regenerative ability, complex tissues and organs do
While regeneration refers to the replacement of a lost body part, the not generally regenerate in response to injury. Can complex tissue
term also describes the repeated replacement of skin cells or shark's regeneration be stimulated in non-regenerating species? These ques-
teeth, the generation of a whole organism from part of an embryo, or in tions are the nexus of regenerative biology and medicine.
the broadest sense, the process of embryogenesis from a fertilized egg The cell is the basic unit of tissue regeneration. Accordingly,
(the regeneration of an organism from a genome). These and other regeneration can occur at multiple levels of biological organization.
examples are classically typed as either physiological (homeostatic) or In one dimension the smallest injury requires repair of a single cell
reparative regeneration, where physiological regeneration describes the (e.g., a severed axon). In another, severe trauma requires replacement
regular replacement of cells and tissues during homeostasis and aging of an entire organ de novo through the coordinated morphogenesis of
(e.g., epidermis, blood, shark's teeth, etc.), and reparative regeneration millions of cells into distinct tissue types (e.g., a limb following
occurs in response to injury (Morgan, 1901). Homeostatic regeneration amputation). Somewhere in between is the regeneration of variably
is a ubiquitous property of vertebrates until that time when cells can no complex structures with multi-cellular architecture and multiple func-
longer replace themselves and tissues and organs begin to fail. When tional units (e.g., ear punch, spinal cord resection, skeletal fracture,
considered in this context, all vertebrates exhibit a capacity for tissue etc.). Regeneration in all of these examples is intimately tied to the
regeneration. The enigma for regeneration biologists is that vertebrate regulated activation, coordinated growth, and differentiation of local
species appear to be distributed along a continuum of reparative cells, be they adult stem cells, or differentiated cells that undergo de-
regenerative ability. As a biological problem, we still do not understand differentiation or reprogramming. How are resident cells activated and
why some adult vertebrates can regenerate organs in response to coordinated to regenerate organs?
damage while others heal similar injuries with scar tissue. Viewed as a Generally speaking, after blood loss is stemmed and an organism


Corresponding authors.
E-mail addresses: awseifert@uky.edu (A.W. Seifert), KMuneoka@cvm.tamu.edu (K. Muneoka).

http://dx.doi.org/10.1016/j.ydbio.2017.08.007
Received 2 June 2017; Received in revised form 28 July 2017; Accepted 4 August 2017
Available online 25 December 2017
0012-1606/ © 2017 Elsevier Inc. All rights reserved.
A.W. Seifert, K. Muneoka Developmental Biology 433 (2018) 190–199

mounts defensive action from infection, local cells accumulate and eration will not occur. Against the backdrop of these features
produce new tissue. Among vertebrates, the process of regenerating a epimorphic regeneration involves two major transformations in re-
replacement organ in situ requires cell proliferation and is referred to sponse to injury: 1) mature tissue into a blastema and 2) a blastema
as epimorphic regeneration (Morgan, 1901). This distinguishes it from into a regenerated organ. Viewed in this light, the blastema is the link
regeneration that involves the reorganization of existing cells at the between healing and morphogenesis.
wound site to restore normal patterning prior to growth (i.e., mor-
phallaxis). Appendage regeneration in salamanders and newts is the 2.1. How to broadly define a blastema?
classic example of a complete and complex epimorphic response (Goss,
1969; Wallace, 1981). While amputating one lobe of the liver will also Blastema: From Greek blastein – to sprout; ma – result of action
stimulate cell (hepatocyte) proliferation to restore organ function, the A blastema is a heterogeneous cell mass that through migration and
lobe itself does not regenerate. What distinguishes epimorphic regen- proliferation transiently forms at the injury site and undergoes
eration from disorganized tissue regeneration is that a regenerating morphogenesis to form the missing organ. We use morphogenesis as
limb or tail forms from a transient proliferative mass called a blastema. it refers to organogenesis (i.e., organ development during embryonic
While a similar response is not usually observed following appendage development), but occurring in the adult form. The heterogeneous
amputation in mammals, there do exist a few mammalian examples of mass of cells is necessarily covered by epidermis (termed the wound
epimorphic regeneration in which a blastema forms, and these can epidermis) and thus the definition of a blastema includes this
instruct the likelihood of enhancing regenerative capabilities in hu- ectoderm-derived component. Historically, a vertebrate blastema was
mans. often defined as a mass of pluripotent cells where blastemal cells would
In this review we focus our attention on digit tip and ear hole contribute to all the of the regenerated structures. In reality, classic and
regeneration as two examples of blastema-based epimorphic regenera- modern lineage tracing techniques have shown that blastemal cells
tion in mammals (Gawriluk et al., 2016; Joseph and Dyson, 1966; tend to respect developmental lineages during normal regeneration
Muneoka et al., 2008). As described below, these two mammalian (rev. in Monaghan and Maden, 2013), although some evidence
models present unique opportunities to discover the mechanisms that suggests that connective tissue fibroblasts over-contribute progeny to
stimulate a regenerative response to injury in lieu of fibrotic healing. In the blastema (Dunis and Namenwirth, 1977; Kragl et al., 2009;
focusing on the digit and ear pinna we do not discuss the broader Muneoka et al., 1986). The blastema has also been classically defined
regenerative abilities of the spiny mice primarily because full-thickness by its developmental potential, i.e., what it transforms into distin-
skin regeneration is not mediated by a blastema (Seifert et al., 2012; guishes the blastema from any other mass of undifferentiated cells
Seifert and Maden, 2014). Similarly, we have chosen to exclude the (e.g., tumor, granulation tissue, etc.). While this may be appropriate
annual shedding and regrowth of deer antlers which represents an during development (nephrogenic blastema) or during salamander
extreme example of physiological regeneration (Kierdorf et al., 2009). limb regeneration (limb blastema) where regeneration routinely oc-
While some authors have suggested deer antler regeneration is curs, this definition is problematic in approaching translational issues
blastema-mediated (Li, 2012) studies have shown that antler regen- associated with regenerative failure. In this regard, it is important to
eration is nerve-independent and the antler bud displays character- modify this classic definition and try to define a blastema by functional
istics that are distinct from an epimorphic blastema (Kierdorf et al., attributes that are essential for establishing its’ developmental poten-
2007, 2009). Thus, while fascinating, in light of these differences and tial, rather than applying a descriptor of its ultimate developmental
the fact that antler regeneration does not occur in response to injury, it fate. A broad definition of this type allows us to identify key properties
is difficult to find parallels with other blastema-mediated regenerative of a blastema that promote the transition from healing to morphogen-
responses in mammals. Ultimately, studying different types of regen- esis and thus provides a basis for comparison across species or across
eration reveals the diversity of regenerative processes. In our view, distinct injury responses within a species. This conceptual framework is
however, understanding the mechanisms that stimulate and regulate particularly useful when examining epimorphic regeneration in mam-
blastema formation is the key to moving from a deconstructive to mals where variation in regenerative ability exists between closely
constructive study of regeneration. related species (ear pinna) and within an individual organ (digit). In
these instances, the presence or absence of a blastema can help provide
2. The vertebrate regeneration blastema an explanation for regenerative success or failure.
In animals that possess regenerative capabilities, local amputation
Several factors have emerged from the vertebrate regeneration initiates the first transformation from mature tissue into a transient
literature as key components in all known examples of blastema-based undifferentiated proliferative phase (blastema) that is followed by the
epimorphic regeneration. These include: (1) formation of a specialized second transformation where morphogenesis and re-differentiation
wound epidermis that functions to attract blastemal cells and maintain replace the missing structures. The first transformation is a specialized
cell proliferation (Globus et al., 1980b; Thornton, 1957a; Thornton and wound healing response that ends with the formation of the blastema
Steen, 1962; Thornton and Thornton 1965), (2) dependence on (Fig. 1). This can be distinguished from a non-regenerative repair
innervation and exposure to nerve or Schwann cell secreted factors response where re-epithelialization is followed by reconstitution of a
(Farkas et al., 2016; Kumar et al., 2007; Mescher et al., 1997; Mullen mature basement membrane, wound contraction, and the deposition of
et al., 1996; Singer, 1952), (3) formation of a pro-regenerative a densely layered fibrous scar tissue that defines regeneration-incom-
extracellular matrix (Calve et al., 2010; Gawriluk et al., 2016; petence. During a regenerative response where the first transformation
Mailman and Dresden, 1979; Marrero et al., 2017; Onda et al., 1991; ends with the formation of a small mass of undifferentiated proliferat-
Satoh et al., 2012; Seifert et al., 2012; Tassava et al., 1996; Vinarsky ing cells, the second transformation involves proliferative expansion of
et al., 2005), (4) deployment of major developmental signaling path- this cell population, patterning, and ultimately the orderly differentia-
ways (rev. in Stoick-Cooper et al., 2007), (5) physical interaction of tion into the multitude of cell types that make up the tissues of the
cells from antonymic positions in three-dimensional space (Carlson, replacement structure. In the context of regenerative failure, these two
1974; Cook and Seifert, 2016; Lheureux, 1975a, 1975b), (6) recogni- transforming events are generally viewed as distinct and independent
tion of uninjured versus new tissue and thus level-specific replacement biological problems. In other words, the study of mammalian regen-
of appropriate tissue to generate a complete organ and (7) a depen- eration involves two distinct and historically separate fields of inquiry:
dence on macrophages to initiate regeneration (Godwin et al., 2013; wound healing, which is largely focused on tissue healing that is non-
Petrie et al., 2014; Simkin et al., 2017). Together, these features regenerative and resolves in the formation of scar tissue, and tissue
contribute to, and support, blastema formation, without which regen- development, which is largely studied during embryogenesis. In

191
A.W. Seifert, K. Muneoka Developmental Biology 433 (2018) 190–199

Fig. 1. Schematic depicting formation of the regenerating mouse digit tip blastema. (A) Cartoon of the mouse digit tip. The P3 bone (blue) is shown as triangular with a proximal base
that contains a bone marrow region (white) and tapers to the distal tip. The bone marrow is highly vascularized (not shown) and hypocellular (red). Epidermis (black) and connective
tissue cells (green) surround the P3 bone. Amputation of the digit tip does not damage the bone marrow. (B) Following amputation of the distal bone the epidermis heals onto the
periosteum of the stump. The distal bone stump contains dead bone (red stars). (C) Histolysis of the stump is mediated by multinucleated osteoclasts (yellow) that erode the bone and
creates a secondary amputation plane. This phase is associated with increased cell numbers in the connective tissue and bone marrow. (D) Once the secondary amputation is complete,
the wound epidermis closes through the region of degraded bone and a blastema forms. Cells from the bone marrow and surrounding connective tissue participate in blastema
formation. The P3 level that regenerates is proximal to the original level of digit amputation.

regeneration competent models, the blastema sits at this interface profiling of cycling tumor cells and blastemal cells may identify a panel
making the study of its formation critical for understanding how of markers specific to cycling blastemal cells. Until these markers are
regeneration is controlled. found, however, one must look to other factors to uniquely define a
Whether or not morphogenesis is an emergent property of the blastema.
blastema or something that must be stimulated is unknown. If the It has long been recognized that a wound epidermis forms atop the
former, then discovering the ability to create a regeneration blastema blastema after re-epithelialization and is required to direct growth of
may be the key to stimulating successful regeneration where it does not the blastema, maintain cell proliferation, and prevent premature
naturally occur (Goss, 1980). Focusing on the first transformation, it differentiation (Globus et al., 1987, 1980b; Thornton, 1960). Studies
remains unresolved if pro-regenerative signals distinct from general- that denude the blastema of the wound epidermis delay regeneration
ized injury signals exist to stimulate blastema formation (Sugiura et al., until re-epithelialization occurs again because the wound epidermis
2016; Tassava and Mescher, 1975). One way or another, the early continually reforms after injury (Thornton, 1957b). In this regard, the
response to injury during epimorphic regeneration involves acute wound epidermis is an integral part of the blastema and essential for
inflammation that is concurrent with re-epithelialization and formation successful regeneration. Once formed, the wound epidermis is further
of the wound epidermis (Gauron et al., 2013; Godwin et al., 2013; Love modified by the action of re-growing axons (Endo et al., 2004; Satoh
et al., 2013; Mescher and Neff, 2005; Simkin et al., 2017). As the et al., 2008). In salamanders and newts, innervation helps transition
inflammatory response resolves, local histolytic activity, re-innerva- the new epidermis into a signaling center called the apical epithelial
tion, and extracellular matrix production facilitates the migration and cap (AEC). This interaction with re-growing nerves is required for
accumulation of cells beneath the wound epidermis to form the nascent blastema formation and distinguishes the AEC from the neoepidermis
blastema (Calve et al., 2010; Singer, 1952; Vinarsky et al., 2005). In that forms atop non-blastema-based regenerating skin wounds (Endo
amphibians and fish, cells of the regenerating limb/fin are derived from et al., 2004; Satoh et al., 2008). Several molecular markers have been
multiple tissue types and lineage restricted progenitor cells have been identified that demarcate this specialized epidermis from epidermis
clearly identified (Knopf et al., 2011; Kragl et al., 2009; Singh et al., located proximal to the injury or from neoepidermis covering tissue
2012; Tu and Johnson, 2011). In addition to the variety of lineage that does not form a blastema (Gawriluk et al., 2016; Han et al., 2001;
restricted progenitor cells that make up the blastema, there is evidence Mullen et al., 1996; Satoh et al., 2008). In addition to molecular
that fibroblastic cells of the interstitial connective tissue are an markers, the wound epidermis is morphologically distinct from the
important cell source for the blastema (Muneoka et al., 1986; Tank normally stratified epithelium typical of adult skin. As regeneration
and Holder, 1979). The connective tissue of the dermis has been progresses reformation of the mature basement membrane beneath the
studied most extensively using cell markers coupled with skin trans- wound epidermis is delayed and its absence is a useful indicator of the
plantation. Such tissue grafting studies show that cells of the dermis wound epidermis (Gawriluk et al., 2016; Neufeld and Day, 1996;
over-contribute to the blastema, and that these cells participate in Neufeld et al., 1996; Seifert et al., 2012). Although further molecular
regenerating skeletal limb tissues (e.g., bone, cartilage, tendons) as well and functional characterization of the wound epidermis is necessary,
as re-forming the dermis (Dunis and Namenwirth, 1977; Kragl et al., current markers and morphological features can identify a wound
2009; Muneoka et al., 1986). Since there are a number of distinct cell epidermis as a fundamental component of the blastema.
types in dermal connective tissue, e.g., vascular, and perivascular cells, Lastly, there is increasing evidence that the blastemal extracellular
and heterogeneity among fibroblasts (Driskell et al., 2013; Rinkevich matrix is a key component of regeneration, although its role as a
et al., 2015) it remains to be demonstrated whether one or all of these facilitator, regulator or passive support structure remains unclear
cell types are multipotent. Nevertheless, since there are no skeletal (Calve et al., 2010; Gawriluk et al., 2016; Mailman and Dresden,
structures in the dermis and yet cells from the dermis can contribute to 1979; Marrero et al., 2017; Onda et al., 1991; Satoh et al., 2012;
regenerated bone and cartilage, the evidence clearly indicates the Seifert et al., 2012; Tassava et al., 1996; Vinarsky et al., 2005). Early
existence of a multipotent cell type within the dermis. Unfortunately, studies of salamander and newt blastemas detected increased levels of
the heterogeneity and plasticity of blastemal cells from tissue-specific matrix proteins associated with cell migration and proliferation (e.g.,
lineages does not aid in the task of defining the blastema generally, fibronectin and tenascin-C) (Onda et al., 1991; Tassava et al., 1996). In
rather, these cellular properties are a function of the specific tissue vitro work examining newt myotubes on fibronectin, tenascin-c and
injured. Instead, a useful definition relies on the fact that some fraction hyaluronic acid showed these proteins could directly regulate cell
of blastemal cells is stimulated to re-enter the cell cycle and undergo behavior (Calve et al., 2010; Calve and Simon, 2012). More recent work
cell cycle progression and division (Globus et al., 1980a; Hay and in mammals (see below) suggests that the extracellular composition of
Fischman, 1961; Tomlinson and Barger, 1987). While the persistence the blastema can itself help define this transient structure when
of active cycling cells can be used to identify the blastema, this alone compared to non-regenerating tissues where a blastema does not form
hardly differentiates a blastema from a tumor. Therefore, the presence (Gawriluk et al., 2016; Marrero et al., 2017; Seifert et al., 2012). All told,
of proliferating cells must be used in combination with other factors. while we have an incomplete definition of a blastema, the indicators
Given that uncontrolled growth is a characteristic of tumor cells and outlined above do provide a conceptual foundation to identify pro-
controlled growth is a characteristic of blastemal cells, comparative regenerative factors from cross-species and inter-injury comparisons.

192
A.W. Seifert, K. Muneoka Developmental Biology 433 (2018) 190–199

3. Two mammalian blastemas – digits and ears stump (Fig. 2H). Wound closure over the stump tissue can be induced
by use of a cyanoacrylic wound dressing, and in this case the bone
As we continue to discover fundamental mechanisms underlying a degradation phase is inhibited and a small blastema forms distal to the
regenerative response to injury, the importance of studying natural original amputation (Simkin et al., 2015b). Alternatively, experimen-
examples of reparative regeneration in mammals is undeniable. Adult tally enhancing the period of bone degradation causes greater stump
regeneration models in mammals, while not as spectacular as limb bone degradation and results in blastemas of a larger size (Sammarco
regeneration in urodeles, do afford researchers a comparative approach et al., 2015). Thus, there appears to be a relationship between the
to understand basic principles of blastema formation and morphogen- extent of bone degradation and blastema size, suggestive of a relation-
esis. To this end, comparing regenerating and non-regenerating ship between tissue histolysis and progenitor cell availability. Blastema
injuries should help expand our ability to define the regeneration maturation is characterized by the onset of skeletal differentiation and
blastema. Moreover, putting phylogenetic considerations aside, impor- progresses in a proximal to distal sequence (Fig. 2H-J). The regenerat-
tant physiological differences between mammals and traditional re- ing new bone forms by direct ossification with osteoblasts secreting
generation models like salamanders, newts and zebrafish (e.g., home- osteoid in a pericellular manner to rapidly form woven bone. The use of
othermy versus poikilothermy, high versus low metabolic rates, etc.) µCT imaging has become an important tool to study the regeneration of
make it vital to understand how tissues can regenerate against the bone during digit tip regeneration. Bone length and volume can be
backdrop of mammalian physiology. As another example, although an tracked and quantitated in vivo and soft tissue changes can be
innate immune response to injury is an ancient weapon against quantitated using contrasting reagents (Fig. 2K-N).
infection, it seems unlikely that this response is exactly the same in The question of how the digit blastema forms is key to under-
newts as it is in a mouse or human. Below we offer two models for standing both regenerative success and regenerative failure, and is an
epimorphic regeneration in mammals where steady progress is con- important step for addressing the question of why most mammals
tributing to our understanding of blastema formation, its definition, studied to date have limited regenerative capabilities. Since a blastema
and more broadly, to the differences between a regenerative or fibrotic can form following digit tip amputation, it is reasonable to ask why a
response to injury. blastema does not form at other amputation levels. The digit blastema
can be defined based on transiently expressed characteristics asso-
3.1. Digit tip regeneration ciated with its formation, and for a more stringent definition, based on
a subset of characteristics that are experimentally shown to be
The regeneration of human fingertips is well documented in the functionally linked to successful regeneration. An approach of this
clinical literature (Illingworth, 1974) and parallels between digit tip kind reflects the general criteria outlined above while also potentially
regeneration in mice and fingertip regeneration in humans have peaked expanding our definition of the blastema. One example is the composi-
interest in the feasibility of human regeneration (Muneoka et al., tion of the extracellular matrix (ECM) produced by blastema cells.
2008). The regenerating mouse digit has become an important experi- Collagen 3 is a minor component of the digit prior to amputation, but
mental model to explore fundamental mechanisms of mammalian becomes a prominent component of the blastema ECM before return-
regeneration and to test strategies aimed at enhancing regenerative ing to baseline levels after differentiation (Marrero et al., 2017; Simkin
capabilities (Dawson et al., 2016). The mouse digit tip regenerates at all et al., 2015b). The cells that produce collagen 3 have been identified as
stages of development, including adults, and the process involves the a subset of digit fibroblasts, called fibroblast reticular cells, and they
formation of a transient blastema. The regeneration competent region display an enhanced proliferative response during blastema formation
consists of the terminal or third phalangeal element (P3), a structurally (Marrero et al., 2017). Collagen 3 production by digit fibroblasts is
unique bone that has the shape of a flattened cone with a wide basal induced in vitro by multi-TNF receptor activation (Katakai et al., 2008;
region that contains a bone marrow cavity and a tapered distal tip Marrero et al., 2017) suggesting that digit fibroblasts specifically react
(Fig. 2A-B). The second phalangeal element (P2) articulates with the to inflammation by producing a provisional matrix that supports a
base of P3 forming the P2/P3 joint (Fig. 2B). The P3 element is encased regenerative response. Although functional studies on the role of
by the nail organ which is required for the regenerative response collagen 3 in digit blastema formation are currently lacking, the data
(Takeo et al., 2013), and a thin layer of loose connective tissue are consistent with the conclusion that a collagen 3 rich matrix
consisting of fibroblasts, vasculature and nerves separate the P3 bone supports mammalian regeneration. A more comprehensive analysis
from the nail epidermis. Amputation through the distal half of P3 of the extracellular environment in regenerative-competent P3 ampu-
initiates the regenerative response, while amputation at the base of P3 tations compared to regeneration-incompetent P2 amputations will
undergoes a healing response with no evidence of regeneration help resolve how matrix proteins contribute to blastema formation.
(Neufeld and Zhao, 1995). After amputation, the continuously growing A number of studies have identified major developmental pathways
nail encases the regenerate, initially appearing as a truncated out- important for digit regeneration through functional analysis. These
growth before eventually conforming to the contour of the blastema include multiple secreted factor signaling pathways including BMP (Yu
(Fig. 2C-E). Internal to the nail the regeneration response involves a et al., 2010), WNT (Lehoczky and Tabin, 2015; Takeo et al., 2013,
series of interconnected and interdependent processes that transforms 2016), PDGF (Johnston et al., 2016), Oncostatin M (Johnston et al.,
the differentiated stump tissues into a blastema that undergoes 2016), CXCL12 (Lee et al., 2013), and VEGF (Yu et al., 2014). Some of
morphogenesis to form replacement tissues. these signaling factors are regulated in the blastema microenvironment
Unlike some other regeneration models, the digit amputation and control cell proliferation, cell migration or cell differentiation
wound does not undergo a rapid re-epithelialization response, instead (Lehoczky, 2016; Simkin et al., 2015a). A series of recent studies have
the epidermis initially heals onto the lateral regions of the amputated identified interactions linking the inflammatory response, angiogen-
stump bone and initiates a histolytic phase (Fig. 2F) that is dominated esis, and re-epithelialization to the control of oxygen availability during
by the recruitment of osteoclasts and the progressive degradation of the the transformation from stump to blastema. Initial studies identifying
stump bone (Fernando et al., 2011; Simkin et al., 2015b). This the central region of the digit blastema as avascular echoed similar
histolytic phase correlates with the acute inflammatory response, and accounts from descriptive studies of the salamander limb blastema
the recruitment of the monocyte/macrophage lineage cells that fuse to (Fernando et al., 2011; Mescher, 1996; Peadon and Singer, 1966; Said
form large multinucleated osteoclasts (Lampiasi et al., 2016) that et al., 2004). The avascular character of the digit blastema correlates
degrade the stump bone (Fig. 2G). Once the stump bone is degraded, with reduced expression of Vegfa and up-regulated expression of the
the epidermis migrates through the region of degraded bone that potent anti-angiogenic factor Pedf, during early regeneration stages in
defines a new amputation level, and the blastema forms distal to the neonates (Muneoka et al., 2008; Yu et al., 2014). Experimental

193
A.W. Seifert, K. Muneoka Developmental Biology 433 (2018) 190–199

Fig. 2. Digit tip regeneration. (A) The distal region of the adult digit tip is comprised of the distal tip of the P3 skeletal element surrounded by a layer of connective tissue and the nail
plate (n). The plane of amputation is indicated by the solid line. (B) Sagittal section through an unamputated P3 element shows the triangle shaped bone (p3) that contains a proximal
bone marrow cavity (bm) and is enclosed by the nail (n). The P3 element articulates with the second phalangeal element (p2) at the P2/P3 joint. The plane of amputation is indicated by
the solid line. (C) External view of a regenerating digit tip at 7 days post-amputation (dpa) showing distal elongation of the nail plate (n) with no remarkable change of the stump bone.
(D) External view of a regenerating digit tip at 12 dpa showing outgrowth of a prominent digit blastema (b) surrounded by the elongating nail (n). (E) External view of a regenerating
digit tip at 17 dpa showing a regenerate that has the general shape of the terminal phalanx. Vasculature associated with ossification is apparent proximal to the distal blastema. (F)
Mallory's triple-stained section of a regenerating digit at 7 dpa showing the absence of wound closure and epidermal attachment to the lateral bone surface. Pitting of the stump bone
surface (p) is observed at this stage. (G) TRAP staining of a 7 dpa regenerate identifies multinucleated osteoclasts (*) associated with the amputated stump bone. (H) Malory's triple-
stained section of a 12 dpa digit showing the distal undifferentiated blastema (b). (I) Mallory's triple-stained regenerate at 17 dpa showing the distal stump with newly differentiated
woven bone (wb) that is capped with undifferentiated blastema cells. (J) At 28 dpa the distal P3 element is regenerated with an interlacing network of new woven bone (wb) that is
histologically distinct from the cortical bone of the stump. (K-M) µCT imaging of regenerating digits stained with a soft-tissue contrast agent (alcoholic iodine) to make the epidermis
radio-opaque and enable analyses of soft tissue versus bone. (K) At 10 dpa the distal cortical bone is eroded and the bone marrow cavity is contiguous with the distal digit blastema (b).
(L) At 17 dpa the regenerated woven bone (wb) appears as a loose network of bony tissue. (M) At 28 dpa the regenerated woven bone (wb) has a mottled appearance and is distinct from
the cortical bone of the stump. (N) Graph showing the normalized longitudinal lengths of the digit tip (solid boxes) and the P3 bone (open boxes) during the regeneration process. The
arrow highlights the transition between bone degradation, blastema formation and osteogenic differentiation of new bone. Both total digit and P3 length regenerates to normal levels
within 3–4 weeks. (O) Hypoxic character of the blastema is shown in a colorimetric overlay of hypoxyprobe staining (pink, O2 < 1.3%) and oxygen-stabilized FBXL5 immunostaining
(green, O2 > 6.0%) as compared to normoxic (purple) areas at 12 dpa. A, C-N from Fernando et al. (2011); B, from Han et al., 2008; O, from Sammarco et al. (2014).

treatment of digit amputations with VEGF induces precocious angio- tissue in full-thickness skin wounds causes excessive re-vascularization
genesis and inhibits skeletal regeneration without inhibiting the that is linked to scar formation (DiPietro, 2016; Wietecha et al., 2015).
formation of a blastema structure, indicating that the avascular nature Thus, the evidence suggests that the regulation of this angiogenesis/
of the blastema is a characteristic essential for regeneration (Yu et al., oxygen/hypoxia cascade is a potential evolutionary target responsible
2014). The region of digit blastema avascularity displays a low oxygen for restricted regenerative capabilities in mammals, and also identifies
tension (< 1.3%) indicating a hypoxic microenvironment (Fig. 2O) and a target pathway to enhance regenerative ability of regeneration
suggestive of a relationship between avascularity and hypoxia incompetent wounds. The extent to which oxygen availability controls
(Sammarco et al., 2014). When oxygen levels are experimentally blastema formation in other examples of epimorphic regeneration
elevated by hyperbaric oxygen treatment, the hypoxic state of the awaits more general investigation.
blastema is eliminated, and this was found to alter the regeneration One of the fundamental questions surrounding epimorphic regen-
response by extending the period of osteoclast-mediated bone degra- eration responses concerns the source and potency of progenitor cells
dation (Sammarco et al., 2015, 2014). While oxygen treatment does not that form the blastema and contribute to the regenerate. As indicated
enhance osteoclastogenesis, it does inhibit the termination of the above, while the identity of these cells may not help define a blastema
osteoclast response, thus extending the period and extent of the bone per se, the source and plasticity of blastemal cells can reveal funda-
histolytic phase. In classic models of bone degradation, osteoclastogen- mental aspects of how cellular phenotypes are modified during
esis is inhibited by production of osteoprotegrin, a secreted decoy regeneration. Lineage tracing studies in mice utilizing cell type specific
receptor that competes with and inhibits RANKL-RANK signaling that promoter-driven reporter expression are a powerful test for multi-
stimulates osteoclastogenesis (Chen et al., 2017). In osteoblasts, potency during regeneration. Not surprisingly, the epidermis has been
osteoprotegrin expression appears directly downstream of Hif2b shown to be lineage restricted during digit tip regeneration as it is
activity which is regulated via the oxygen sensing activity of prolyl during salamander limb regeneration (Kragl et al., 2009; Rinkevich
hydroxylase enzymes (Wu et al., 2015). These studies identify a et al., 2011), and cell labeling studies show that cells derived from
dynamic process that involves regulating angiogenesis to establish a hematopoietic stem cells do not contribute to the major structural
transient hypoxic blastema microenvironment that functions as a tissues of the regenerated digit (Rinkevich et al., 2011). Endothelial
regulatory switch to transition the histolytic phase prior to blastema cells and osteoblasts contribute to the blastema and regenerated digit,
formation to the growth phase once the blastema has formed. It is and both are lineage-restricted with regard to their potency (Lehoczky
interesting that the pro-angiogenic microenvironment of granulation et al., 2011; Rinkevich et al., 2011). The use of promoter-specific Cre

194
A.W. Seifert, K. Muneoka Developmental Biology 433 (2018) 190–199

expression coupled with a constitutively active reporter gene to track Dyson, 1966). Cell cycle re-entry, progression and proliferation are
cell lineage has also been useful in determining whether specific cell observed in blastemal cells and a small group of epidermal cells at the
types change phenotype during regeneration. Such studies show that proximal wound margins (Gawriluk et al., 2016; Seifert et al., 2012). As
Sox9-expressing skeletal cells, Scx-expressing tendon cells, and Tie2- the ear hole is progressively filled, new tissue growth preferentially
expressing endothelial cells do not undergo transdifferentiation during occurs in the proximal portion of the hole (Fig. 3C-C′) and new tissue
regeneration (Rinkevich et al., 2011). While lineage studies have led to growth is coincident with axon regeneration (Gawriluk et al., 2016) and
the general conclusion that the digit blastema is composed of only angiogenesis (Joseph and Dyson, 1966; Matias et al., 2015). Re-
lineage restricted cells (Lehoczky et al., 2011; Rinkevich et al., 2011), differentiation of new tissues occurs outside to inside such that new
this generalized conclusion must be weighed against the limited hair follicles and cartilage form nearer to the wound margin before they
number of cell types that have been studied, and that the cell types appear in the center (Fig. 3C-D′) (Joseph and Dyson, 1966; Seifert
contributing to the regenerated digit (epidermis, endothelial cells, et al., 2012). Hair follicle regeneration in spiny mice is visible twenty
osteoblasts) are lineage restricted during development. Indeed, with days post injury. As new tissue forms to close spiny mouse ear holes,
the exception of hematopoietic stem cells that do not contribute to elastic cartilage regeneration occurs at two positions. Similar to
regenerated structures, the contribution of more phenotypically labile fracture healing, a callous forms at the cut end of the cartilage sheet
cells such as mesenchymal stem cells or tissue-specific stem cells and this appears to contribute some portion of the new cartilage.
remain unknown. Current studies show that a panel of positive and However, the majority of the new cartilage appears to emerge de novo
negative markers is required to identify certain multipotent progenitor from mesenchymal condensations within the new tissue (Seifert et al.,
cells making it difficult to locate and trace these cells in vivo (Caplan 2012). Eventually, new tissue completely fills the hole and cellular
and Correa, 2011; Gökçinar-Yagci et al., 2015). Nevertheless, currently differentiation restores the excised tissue. These interconnected and
available evidence indicates that a number of cell types participating in overlapping processes together constitute regeneration of a complex
digit regeneration are lineage restricted. musculoskeletal structure. Importantly, provided holes larger than
2 mm are made through the pinna, epimorphic regeneration of ear
3.2. Ear pinna regeneration holes can be differentiated from fibrosis-driven partial ear hole closure
observed in other rodents and laboratory mouse strains (Gawriluk
The regeneration of holes made through the ear pinna has long et al., 2016).
been recognized as another example of epimorphic regeneration in Whereas the digit tip provides a model to study regenerative
mammals (Goss and Grimes, 1972; Vorontsova and Liosner, 1960). success and failure in the same organ (e.g., successful regeneration in
The external ear pinna is a complex organ comprised of skin, adipose P3 versus regenerative failure in P2), the ear hole model of epimorphic
tissue, skeletal muscle, vasculature, nerves, and elastic cartilage regeneration exploits interspecies variation in regenerative ability. In
(Fig. 3A-B). In some species, a complete hole punched through the contrast to regeneration observed in rabbits and spiny mice, broad
pinna removes portions of all of these structures and initiates a phylogenetic sampling shows that the ability to regenerate ear holes is
regenerative response (Fig. 3C-E). The phenomenon was first reported restricted among mammals (Gawriluk et al., 2016; Goss, 1980;
in 1953 with the observation that 1 cm holes made in rabbit ears Williams-Boyce and Daniel, 1986). Ear hole regeneration may extend
completely closed with new tissue including new hair follicles, skin and to chinchillas, pikas and cats, but other rodents so far studied are
cartilage (Markelova cited by Vorontsova and Liosner (1960)). Ensuing unable to regenerate ear holes. This raises the question as to how
investigations in the rabbit ear extended these observations and regeneration occurs in some mammalian species? With the discovery of
provided evidence that the type of epidermis (i.e., abdominal vs. ear) regeneration in spiny mice, one approach has been to compare the
and presence of auricular cartilage were required for successful regenerative response of the ear pinna to fibrotic repair of the same
regeneration (Goss and Grimes, 1972; Grimes and Goss, 1972; injury in other rodents. Comparing spiny mice to outbred Mus
Joseph and Dyson, 1966; Williams-Boyce and Daniel, 1980). musculus showed that formation and persistence of a blastema during
Interestingly, semicircular wounds or notches made at the edges of healing distinguishes the regenerative response from a scarring re-
the ear were reported to initiate, but not complete regeneration sponse (Gawriluk et al., 2016; Seifert et al., 2012). This distinction
(Williams-Boyce and Daniel, 1980). More recently, work with two provides an opportunity to characterize a mammalian blastema and
species of wild African spiny mouse (Acomys kempi and A. percivali) tease apart cellular and molecular processes that operate during these
led to the discovery that these rodents could regenerate 4 mm holes in differential responses to injury.
the ear pinna (Seifert et al., 2012) and subsequent research extended Careful analysis of tissue regeneration in spiny mice has begun to
these findings to include another species, A. cahirinus (Gawriluk et al., help define cellular and molecular features of the ear blastema
2016; Matias et al., 2015). Comprehensive studies in spiny mice and (Gawriluk et al., 2016; Matias et al., 2015; Seifert et al., 2012;
rabbits have detailed the regeneration of full-thickness skin, hair Simkin et al., 2017). As indicated above, a key characteristic of the
follicles, glands, adipose tissue, elastic cartilage, and to a limited blastema is its epidermal compartment, the wound epidermis, which
degree, muscle fibers (Fig. 3E) (Gawriluk et al., 2016; Joseph and forms following re-epithelialization. As mesenchymal cells accumulate
Dyson, 1966; Matias et al., 2015). That some species can regenerate ear beneath the epidermis distal to the cut cartilage, epidermal cell
holes while others cannot make this model of epimorphic regeneration proliferation is excluded from the wound epidermis and maintained
attractive to uncover mechanisms that stimulate blastema formation in a small group of wound edge keratinocytes (Gawriluk et al., 2016).
and differentiate a regenerative or fibrotic response to injury. Basal keratinocytes in this distal epidermis maintain a circular
Superficially, the regenerative response follows the stereotypical morphology rather than assume the columnar morphology typical of
pattern observed during other examples of vertebrate appendage normal stratified epidermis and the distal epidermis observed in mice
regeneration. The initial trauma stimulates an injury response that during scarring. Importantly, keratin 17 is upregulated in the wound
recruits neutrophils and monocytes to the injury site, while the epidermis and expressed until the ear hole is filled with new tissue, a
hemostatic response contributes to scab formation (Simkin et al., situation that does not occur during scarring where keratin 17
2017). Shortly thereafter, wound edge keratinocytes are stimulated to expression disappears by D15 post injury (Gawriluk et al., 2016).
migrate beneath the scab and re-epithelialize the injury (Gawriluk These features are coincident with a failure to reconstitute the lamina
et al., 2016; Joseph and Dyson, 1966; Seifert et al., 2012). Once the densa beneath the distal epidermis until hole closure is complete
epidermal barrier is restored, cells begin to accumulate centripetally (Gawriluk et al., 2016; Seifert et al., 2012). Together, these features
beneath the neoepidermis and as this accumulation continues a characterize the unique epidermal compartment of the blastema which
regeneration blastema forms (Gawriluk et al., 2016; Joseph and persists until hole closure is completed and the circular blastema

195
A.W. Seifert, K. Muneoka Developmental Biology 433 (2018) 190–199

Fig. 3. 4 mm ear punch assay and epimorphic regeneration in the spiny mouse Acomys cahirinus. (A) Position of a normal ear punch in an adult spiny mouse relative to the ear pinna
along the proximodistal (p/d) axis. (B) Masson's Trichrome stained tissue section of an uninjured spiny mouse ear pinna indicating: epidermis (e), dermis (d), adipose tissue (ad),
skeletal muscle (m), and elastic cartilage (c). Hair follicles and sebaceous glands are visible in the dorsal and ventral skin. Scale bar = 50 µm. (C-D) Whole mount images of regenerating
ear holes at D30 (C) and D40 (D) post injury along the proximodistal (p/d) axis. In regenerating ears growth is biased to the proximal part of the hole. White circles indicate original
4 mm punch. (C′-D′) Sections through C and D showing the regeneration blastema (C′) and ear hole closure (D′). New hair follicles, dermis, adipose tissue and cartilage are visible at
D40. Cartoon at right in C′ shows how tissue sections are collected. (E) A regenerated ear hole in Acomys cahirinus at D85 showing complete regeneration along the proximodistal and
dorsoventral axes. New muscle fibers are visible past the proximal amputation plane. Dotted black lines indicate the injury place (C′-E). Image in E modified slightly from Gawriluk et al.
(2016).

disappears. Further molecular characterization of the wound epidermis ECM in spiny mice, work across regenerating vertebrates strongly
in spiny mice and other vertebrates will help refine our understanding supports ECM composition as a key marker of a regeneration blastema
of how this compartment regulates blastema formation and regenera- (Calve et al., 2010; Gawriluk et al., 2016; Marrero et al., 2017; Onda
tion. et al., 1991; Satoh et al., 2012; Seifert et al., 2012; Tassava et al., 1996).
Cells accumulating beneath the wound epidermis constitute the While comparative transcriptomics may suggest that the blastema
mesenchymal compartment of the blastema, and another salient extracellular environment has a unique molecular signature
feature of the blastema is the unique composition of the extracellular (Gawriluk et al., 2016), the extent to which known and novel matrix
matrix (ECM) that supports specific activities of blastemal cells during molecules are functionally required for blastema formation and
morphogenesis. Fibrotic ECM is dominated by collagen with a ratio morphogenesis awaits testing.
favoring collagen 1 > collagen 3 (Gawriluk et al., 2016; Lo et al., 2012; Epimorphic regeneration in an adult occurs in the context of a fully
Marrero et al., 2017; Seifert and Maden, 2014; Simkin et al., 2015b). differentiated and operational immune system. Prior to blastema
Analysis of blastema ECM in comparison to matrix composition during formation, local injury elicits an immune response. Although innate
fibrotic healing revealed a profound difference in the extracellular and adaptive immune defenses occur in both scarring and regenerating
environment (Gawriluk et al., 2016; Seifert et al., 2012). While collagen systems, recent work from mammals and other vertebrates suggests
1 was produced during mammalian regeneration, it comprised only a that aspects of these responses may positively regulate a regenerative
small fraction of the blastemal ECM, which was instead dominated by response (Gauron et al., 2013; Godwin et al., 2013; Lai et al., 2017;
fibronectin and tenascin-C among other matrix proteins (Gawriluk Love et al., 2013; Petrie et al., 2014; Simkin et al., 2017). Comparing
et al., 2016). Fibronectin and tenascin-C promote cell migration and the inflammatory response during regeneration and scarring in spiny
proliferation, two cellular attributes required for morphogenesis during mice revealed that reactive oxygen species produced by NAPDH-
regeneration. Beyond the capacity of ECM proteins to physically oxidase at the injury site were significantly elevated during regenera-
interact with cells and stimulate behavior, the ECM also acts as a tion and these molecules persisted in elevated amounts during
reservoir for growth factors, cytokines and signaling molecules. blastema formation (Simkin et al., 2017). This mirrors a proposed role
Although more work is required to fully characterize the blastemal for reactive oxygen species during tail regeneration in Xenopus and

196
A.W. Seifert, K. Muneoka Developmental Biology 433 (2018) 190–199

zebrafish (Gauron et al., 2013; Love et al., 2013) and brain regenera- 4. Conclusions: expanding the limits of regeneration
tion in newts (Hameed et al., 2015). Macrophages are a key regulator of
the local immune response and vertebrate studies suggests that they The continued study of epimorphic regeneration in mammals has
control inflammation, fibrosis and blastema formation during regen- much to add to our understanding of vertebrate regeneration
eration (Godwin et al., 2013; Petrie et al., 2014; Simkin et al., 2017). biology. The ear hole model discussed above provides an experi-
Depleting macrophages during normal fibrotic wound healing demon- mental system to uncover key mechanisms that explain differences
strated they control the rate of re-epithelialization, fibrosis and cell in regenerative ability across mammalian species. When applied
proliferation (Leibovich and Ross, 1975; Mirza et al., 2009). Similarly, more broadly, it may also help identify additional mammalian
macrophages infiltrate the ear pinna and are present during regenera- species that possess enhanced regenerative ability. As a comparative
tion and scarring. Depleting macrophages in the ear pinna prior to and system between regenerating and non-regenerating species, the ear
during injury demonstrated a functional requirement for these cells punch assay will reveal whether specific molecules or pathways can
during ear hole regeneration (Simkin et al., 2017). Interestingly, serve as early indicators for blastema formation and which factors
depleting macrophages delayed re-epithelialization and inhibited blas- alone or in combination are required to maintain blastema morpho-
tema formation, and when macrophages were allowed to infiltrate the genesis. This will be necessary if we hope to uncover potential
ear pinna a blastema formed and regeneration ensued. Beyond the targets that might be used to stimulate regeneration. In this light,
assessment that macrophages are generally required for epimorphic both the ear hole and digit tip models offer an added translational
regeneration, some evidence suggests specific macrophage subtypes perspective to the biology of regeneration. In digit studies, amputa-
may differentially regulate this response to injury (Mescher, 2017; tion of the mouse P2 digit has become a useful regeneration
Simkin et al., 2017). Interestingly, when macrophage subtypes were incompetent model to test strategies for inducing a regeneration
analyzed during spiny mouse ear hole regeneration, classically acti- response (Dawson et al., 2016). BMP signaling has been shown to be
vated (M1) macrophages were largely absent from the blastema important for digit tip regeneration (Yu et al., 2010) and digit
(Simkin et al., 2017). This study left open whether the blastema amputation at a P2 level is stimulated to regenerate a patterned
triggered macrophage phenotype switching or instead certain subtypes response by targeted BMP2 treatment in both neonates and adult
were restricted from infiltrating the blastema. Further studies will need mice (Dawson et al., 2017; Yu et al., 2012). The BMP2 response
to determine the importance of particular macrophage subtypes and indirectly induces the recruitment of cells to the amputation wound
the extent to which these subtypes may regulate blastema formation by activating CXCL12 production (Lee et al., 2013) and BMP2
either by directly stimulating local fibroblasts or indirectly by polariz- functions as a mitogen for chondrocytes that establish an endochon-
ing the immune response. dral ossification center at the amputation wound (Dawson et al.,
The identity and plasticity of progenitor cells that form the ear 2017; Yu et al., 2012). Once formed, the induced endochondral
blastema is currently unknown. The mesenchymal compartment of the ossification center organizes the morphogenesis of new distal
ear pinna develops entirely from Hoxa2-expressing cranial neural crest of skeletal tissue that replaces the amputated P2 bone. These studies
the second pharyngeal arch, whereas the epidermal compartment is provide proof of concept that a regenerative response can be
derived from ectoderm (Minoux et al., 2013). Whereas subsequent stimulated at a regeneration incompetent amputation wound, and
differentiation of these neural crest progenitors during external ear when combined with results from the ear leads to two important
development is less well understood, Hoxa2 regulates BMP signaling, conclusions. First, progenitor cell availability is not a limiting factor
specifically Bmp5 and Bmp4, during cartilage development in the pinna at a mammalian amputation injury. Indeed, it seems unlikely that
(Minoux et al., 2013). While the prevailing view of the cellular contribu- different progenitor cell populations would be present in the second
tion to the blastema is that lineage restricted cells will supply progenitors and third phalanx of the digit or in the ear pinna from different
to a heterogeneous pool that will expand and differentiate into specific rodent species. Second, limitation of the wound microenvironment
tissue subsets, what remains unknown is the degree to which lineage to activate progenitor cells is in part responsible for regenerative
restriction applies to local cells that contribute to the ear blastema. For failure. How exactly this activation is controlled remains the focus of
instance, if the shared cranial crest cell origin of all non-epidermal tissue current and future investigations. As studies using mammalian
structures in the ear pinna sets the plasticity limit during regeneration, regeneration models progress they will continue providing insight
then in theory, the pool of available cells for regeneration would extend to into how injured tissue can be transformed into a regeneration
all mesenchymal cells of the ear. Similarly, if regeneration in the ear is blastema and how blastemal morphogenesis is controlled to appro-
dependent on reserves of adult stem cells rather than some form of de- priately replace injured tissue in situ. This will require dissecting
differentiation then fewer populations would be necessary to replace all components of the blastema to uncover new characteristics or
the missing tissues in the ear. A different question is whether the markers that can indicate when these two transformations take
appropriate cells are present for regeneration, but they are simply unable place and using these markers to test approaches at successfully
to form a blastema. In this scenario, heterogeneous blastemal cells that stimulating regeneration where it does not normally occur.
normally contribute to regeneration in spiny mice are resistant to cell
cycle re-entry, progression and division in non-regenerating mammals.
Acknowledgements
Evidence to support the latter comes from research showing that while the
number of Ki67+ cells in the ear during regeneration and scarring is
We would like to thank Jennifer Simkin and two anonymous
similar, cell cycle progression and division is rarely observed during
reviewers for comments and critical reading of the manuscript and
scarring (Gawriluk et al., 2016). This result was partly attributed to the
Ricardo Zayas and Karen Echeverri for soliciting our review. A.W.
observation that p21 and p27 were found throughout cell nuclei during
Seifert is supported by the National Science Foundation (NSF) and the
scarring, but were absent from the blastema during regeneration.
Office for International Science and Engineering (OISE) (IOS
Importantly, the blastema in spiny mice was significantly enriched with
-1353713) and the National Institute of Arthritis and
markers for the G1/S transition (pRb), DNA replication (EdU) and
Musculoskeletal and Skin Diseases (NIAMS – NIH) (R01AR070313).
mitosis (PHH3) which differentiated blastema cells from cells present
K. Muneoka is supported by Texas A & M University.
during fibrotic healing in lab mice (Gawriluk et al., 2016). Understanding
whether or not cell proliferation occurs in response to specific paracrine
signals or results from an intrinsic ability to undergo cell cycle progression Competing interests
will be vital in discovering how spiny mice form an ear blastema and
undergo regeneration. The authors declare no competing interests.

197
A.W. Seifert, K. Muneoka Developmental Biology 433 (2018) 190–199

References Kierdorf, U., Li, C., Price, J.S., 2009. Improbable appendages: deer antler renewal as a
unique case of mammalian regeneration. Semin. Cell Dev. Biol. 20, 535–542.
Knopf, F., Hammond, C., Chekuru, A., Kurth, T., Hans, S., Weber, C.W., Mahatma, G.,
Calve, S., Odelberg, S.J., Simon, H.G., 2010. A transitional extracellular matrix instructs Fisher, S., Brand, M., Schulte-Merker, S., Weidinger, G., 2011. Bone regenerates via
cell behavior during muscle regeneration. Dev. Biol. 344, 259–271. dedifferentiation of osteoblasts in the zebrafish fin. Dev. Cell 20, 713–724.
Calve, S., Simon, H.-G., 2012. Biochemical and mechanical environment cooperatively Kragl, M., Knapp, D., Nacu, E., Khattak, S., Maden, M., Epperlein, H.H., Tanaka, E.M.,
regulate skeletal muscle regeneration. FASEB J. 26, 2538–2545. 2009. Cells keep a memory of their tissue origin during axolotl limb regeneration.
Caplan, A.I., Correa, D., 2011. The MSC: an injury drugstore. Cell stem Cell 9, 11–15. Nature 460, 60–65.
Carlson, B.M., 1974. Morphogenetic interactions between rotated skin cuffs and Kumar, A., Godwin, J.W., Gates, P.B., Garza-Garcia, A.A., Brockes, J.P., 2007. Molecular
underlying stump tissues in regenerating axolotl forelimbs. Dev. Biol. 39, 263–285. basis for the nerve dependence of limb regeneration in an adult vertebrate. Science
Chen, X., Wang, Z., Duan, N., Zhu, G., Schwarz, E.M., Xie, C., 2017. Osteoblast-osteoclast 318, 772–777.
interactions. Connect Tissue Res., 1–9. Lai, S.L., Marin-Juez, R., Moura, P.L., Kuenne, C., Lai, J.K.H., Tsedeke, A.T., Guenther,
Cook, A.B., Seifert, A.W., 2016. Beryllium nitrate inhibits fibroblast migration to disrupt S., Looso, M., Stainier, D.Y., 2017. Reciprocal analyses in zebrafish and medaka
epimorphic regeneration. Development 143, 3491–3505. reveal that harnessing the immune response promotes cardiac regeneration. Elife, 6.
Dawson, L.A., Simkin, J., Sauque, M., Pela, M., Palkowski, T., Muneoka, K., 2016. Lampiasi, N., Russo, R., Zito, F., 2016. The alternative faces of macrophage generate
Analogous cellular contribution and healing mechanisms following digit amputaton osteoclasts. Biomed. Res. Int. 2016, 9089610.
and phalangeal fracture in mice. Regeneration 3, 13. Lee, J., Marrero, L., Yu, L., Dawson, L.A., Muneoka, K., Han, M., 2013. SDF-1alpha/
Dawson, L.A., Yu, L., Yan, M., Marrero, L., Schanes, P.P., Dolan, C., Pela, M., Peterson, CXCR4 signaling mediates digit tip regeneration promoted by BMP-2. Dev. Biol. 382,
B., Han, M., Muneoka, K., 2017. The periosteal requirement and temporal dynamics 98–109.
of BMP2-induced middle phalanx regeneration in the adult mouse. Regeneration, 4. Lehoczky, J.A., 2016. Are fingernails a key to unlocking the puzzle of mammalian limb
DiPietro, L.A., 2016. Angiogenesis and wound repair: when enough is enough. J. Leukoc. regeneration? Exp. Dermatol..
Biol. 100, 979–984. Lehoczky, J.A., Robert, B., Tabin, C.J., 2011. Mouse digit tip regeneration is mediated by
Driskell, R.R., Lichtenberger, B.M., Hoste, E., Kretzschmar, K., Simons, B.D., fate-restricted progenitor cells. Proc. Natl. Acad. Sci. USA 108, 20609–20614.
Charalambous, M., Ferron, S.R., Herault, Y., Pavlovic, G., Ferguson-Smith, A.C., Lehoczky, J.A., Tabin, C.J., 2015. Lgr6 marks nail stem cells and is required for digit tip
Watt, F.M., 2013. Distinct fibroblast lineages determine dermal architecture in skin regeneration. Proc. Natl. Acad. Sci. USA 112, 13249–13254.
development and repair. Nature 504, 277–281. Leibovich, S.J., Ross, R., 1975. The role of the macrophage in wound repair. A study with
Dunis, D.A., Namenwirth, M., 1977. The role of grafted skin in the regeneration of x- hydrocortisone and antimacrophage serum. Am. J. Pathol. 78, 71–100.
irradiated axolotl limbs. Dev. Biol. 56, 97–109. Lheureux, E., 1975a. Nouvelles donnees sur les roles de la peau et des tissues internes
Endo, T., Bryant, S.V., Gardiner, D.M., 2004. A stepwise model system for limb dans la regeneration du membre du triton Pleurodeles waltlii, Michah (amphibian
regeneration. Dev. Biol. 270, 135–145. urodele). Wilhelm. Roux. Arch. 776, 285–301.
Farkas, J.E., Freitas, P.D., Bryant, D.M., Whited, J.L., Monaghan, J.R., 2016. Lheureux, E., 1975b. Régénération des membres irradiés dePleurodeles waltlii Michah.
Neuregulin-1 signaling is essential for nerve-dependent axolotl limb regeneration. (Urodčle). Influence des qualités et orientations des greffons non irradies. Wilhelm.
Development 143, 2724–2731. Roux'Arch. Entwickl. Org. 176, 303–327.
Fernando, W.A., Leininger, E., Simkin, J., Li, N., Malcom, C.A., Sathyamoorthi, S., Han, Li, C., 2012. Deer antler regeneration: a stem cell-based epimorphic process. Birth
M., Muneoka, K., 2011. Wound healing and blastema formation in regenerating digit Defects Res. C Embryo Today 96, 51–62.
tips of adult mice. Dev. Biol. 350, 301–310. Lo, D.D., Zimmermann, A.S., Nauta, A., Longaker, M.T., Lorenz, H.P., 2012. Scarless
Gauron, C., Rampon, C., Bouzaffour, M., Ipendey, E., Teillon, J., Volovitch, M., Vriz, S., fetal skin wound healing update. Birth Defects Res. Part C: Embryo Today: Rev. 96,
2013. Sustained production of ROS triggers compensatory proliferation and is 237–247.
required for regeneration to proceed. Sci. Rep. 3, 2084. Love, N.R., Chen, Y., Ishibashi, S., Kritsiligkou, P., Lea, R., Koh, Y., Gallop, J.L., Dorey,
Gawriluk, T.R., Simkin, J., Thompson, K.L., Biswas, S.K., Clare-Salzler, Z., Kimani, J.M., K., Amaya, E., 2013. Amputation-induced reactive oxygen species are required for
Kiama, S.G., Smith, J.J., Ezenwa, V.O., Seifert, A.W., 2016. Comparative analysis of successful Xenopus tadpole tail regeneration. Nat. Cell Biol. 15, 222–228.
ear-hole closure identifies epimorphic regeneration as a discrete trait in mammals. Mailman, M.L., Dresden, M.H., 1979. Denervation effects on newt limb regeneration:
Nat. Commun. 7, 11164. collagen and collagenase. Dev. Biol. 71, 60–70.
Globus, M., Vethamany-Globus, S., Kesik, A., 1987. Control of blastema cell proliferation Marrero, L., Simkin, J., Sammarco, M.C., Muneoka, K., 2017. Fibroblast reticular cells
by possible interplay of calcium and cyclic nucleotides during newt limb engineer a blastema extracellular network during digit tip regeneration in mice.
regeneration. Differentiation 35, 94–99. Regeneration 4, 16.
Globus, M., Vethamany-Globus, S., Lee, Y.C., 1980a. Effect of apical epidermal cap on Matias, D.S., Martins, A.R., Casanellas, I., Ova, A.B., Araújo, I.M., Power, D., Tiscornia,
mitotic cycle and cartilage differentiation in regeneration blastemata in the newt, G., 2015. Ear wound regeneration in the African spiny mouse Acomys cahirinus.
Notophthalmus viridescens. Dev. Biol. 75, 358–372. Regeneration.
Globus, M., Vethamany-Globus, S., Lee, Y.C.I., 1980b. Effect of apical epidermal cap on Mescher, A.L., 1996. The cellular basis of limb regeneration in urodeles. Int. J. Dev. Biol.
mitotic cycle and cartilage differentiation in regeneration blastemata in the newt, 40, 785–795.
Notophthalmus viridescens. Dev. Biol. 75, 358–372. Mescher, A.L., 2017. Macrophages and fibroblasts during inflammation and tissue repair
Godwin, J.W., Pinto, A.R., Rosenthal, N.A., 2013. Macrophages are required for adult in models of organ regeneration. Regeneration, (n/a-n/a).
salamander limb regeneration. Proc. Natl. Acad. Sci. USA 110, 9415–9420. Mescher, A.L., Connell, E., Hsu, C., Patel, C., Overton, B., 1997. Transferrin is necessary
Gökçinar-Yagci, B., Uçkan-Çetinkaya, D., Çelebi-Saltik, B., 2015. Pericytes: properties, and sufficient for the neural effect on growth in amphibian limb regeneration
functions and applications in tissue engineering. Stem Cell Rev. Rep. 11, 549–559. blastemas. Dev. Growth Differ. 39, 677–684.
Goss, R.J., 1969. Principles of Regeneration. Academic Press, New York. Mescher, A.L., Neff, A.W., 2005. Regenerative capacity and the developing immune
Goss, R.J., 1980. Prospects for regeneration in man. Clin. Orthop. Relat. Res. 151, system. Adv. Biochem. Eng. Biotechnol. 93, 39–66.
270–282. Minoux, M., Kratochwil, C.F., Ducret, S., Amin, S., Kitazawa, T., Kurihara, H., Bobola, N.,
Goss, R.J., Grimes, L.N., 1972. Tissue interactions in regeneration of rabbit ear holes. Vilain, N., Rijli, F.M., 2013. Mouse Hoxa2 mutations provide a model for microtia
Am. Zool. 12, 151–157. and auricle duplication. Development 140, 4386–4397.
Grimes, L.N., Goss, R.J., 1972. Morphogenetic influence of cartilage in regenerating Mirza, R., DiPietro, L.A., Koh, T.J., 2009. Selective and specific macrophage ablation is
rabbit ear holes. Am. Zool. 12, 708–709. detrimental to wound healing in mice. Am. J. Pathol. 175, 2454–2462.
Hameed, L.S., Berg, D.A., Belnoue, L., Jensen, L.D., Cao, Y., Simon, A., 2015. Monaghan, J.R., Maden, M., 2013. Cellular plasticity during vertebrate appendage
Environmental changes in oxygen tension reveal ROS-dependent neurogenesis and regeneration. Curr. Top. Microbiol. Immunol. 367, 53–74.
regeneration in the adult newt brain. Elife, 4. Morgan, T.H., 1901. Regeneration. The Macmillan Company; Macmillan & Co., ltd., New
Han, M.J., An, J.Y., Kim, W.S., 2001. Expression patterns of Fgf-8 during development York, London.
and limb regeneration of the axolotl. Dev. Dyn. 220, 40–48. Mullen, L.M., Bryant, S.V., Torok, M.A., Blumberg, B., Gardiner, D.M., 1996. Nerve
Han, M., Yang, X., Lee, J., Allan, C.H., Muneoka, K., 2008. Development and dependency of regeneration: the role of distal-less and FGF signaling in amphibian
regeneration of the neonatal digit tip in mice. Dev. Biol. 315, 125–135. limb regeneration. Development 122, 3487–3497.
Hay, E.D., Fischman, D.A., 1961. Origin of the blastema in regenerating limbs of the Muneoka, K., Allan, C.H., Yang, X., Lee, J., Han, M., 2008. Mammalian regeneration and
newt Triturus viridescens. An autoradiographic study using tritiated thymidine to regenerative medicine. Birth Defects Res. C Embryo Today 84, 265–280.
follow cell proliferation and migration. Dev. Biol. 3, 26–59. Muneoka, K., Fox, W.F., Bryant, S.V., 1986. Cellular contribution from dermis and
Illingworth, C.M., 1974. Trapped fingers and amputated finger tips in children. J. cartilage to the regenerating limb blastema in axolotls. Dev. Biol. 116, 256–260.
Pediatr. Surg. 9, 853–858. Neufeld, D.A., Day, F.A., 1996. Perspective: a suggested role for basement membrane
Johnston, A.P., Yuzwa, S.A., Carr, M.J., Mahmud, N., Storer, M.A., Krause, M.P., Jones, structures during newt limb regeneration. Anat. Rec. 246, 155–161.
K., Paul, S., Kaplan, D.R., Miller, F.D., 2016. Dedifferentiated schwann cell Neufeld, D.A., Day, F.A., Settles, H.E., 1996. Stabilizing role of the basement membrane
precursors secreting paracrine factors are required for regeneration of the and dermal fibers during newt limb regeneration. Anat. Rec. 245, 122–127.
mammalian digit tip. Cell Stem Cell 19, 433–448. Neufeld, D.A., Zhao, W., 1995. Bone regrowth after digit tip amputation in mice is
Joseph, J., Dyson, M., 1966. Tissue replacement in the rabbit's ear. Br. J. Surg. 53, equivalent in adults and neonates. Wound Repair Regen. 3, 461–466.
372–380. Onda, H., Poulin, M.L., Tassava, R.A., Chiu, I.M., 1991. Characterization of a newt
Katakai, T., Suto, H., Sugai, M., Gonda, H., Togawa, A., Suematsu, S., Ebisuno, Y., tenascin cDNA and localization of tenascin mRNA during newt limb regeneration by
Katagiri, K., Kinashi, T., Shimizu, A., 2008. Organizer-like reticular stromal cell layer in situ hybridization. Dev. Biol. 148, 219–232.
common to adult secondary lymphoid organs. J. Immunol. 181, 6189–6200. Peadon, A.M., Singer, M., 1966. The blood vessels of the regenerating limb of the adult
Kierdorf, U., Kierdorf, H., Szuwart, T., 2007. Deer antler regeneration: cells, concepts, newt, Triturus. J. Morphol. 118, 79–89.
and controversies. J. Morphol. 268, 726–738. Petrie, T.A., Strand, N.S., Yang, C.T., Rabinowitz, J.S., Moon, R.T., 2014. Macrophages

198
A.W. Seifert, K. Muneoka Developmental Biology 433 (2018) 190–199

modulate adult zebrafish tail fin regeneration. Development 141, 2581–2591. Takeo, M., Hale, C.S., Ito, M., 2016. Epithelium-derived Wnt ligands are essential for
Rinkevich, Y., Lindau, P., Ueno, H., Longaker, M.T., Weissman, I.L., 2011. Germ-layer maintenance of underlying digit bone. J. Investig. Dermatol. 136, 1355–1363.
and lineage-restricted stem/progenitors regenerate the mouse digit tip. Nature 476, Tank, P.W., Holder, N., 1979. The distribution of cells in the upper forelimb of the
409–413. axolotl, Ambystoma mexicanum. J. Exp. Zool. 209, 435–442.
Rinkevich, Y., Walmsley, G.G., Hu, M.S., Maan, Z.N., Newman, A.M., Drukker, M., Tassava, R., Mescher, A., 1975. The roles of injury, nerves, and the wound epidermis
Januszyk, M., Krampitz, G.W., Gurtner, G.C., Lorenz, H.P., Weissman, I.L., during the initiation of amphibian limb regeneration. Differentiation 4, 23–24.
Longaker, M.T., 2015. Skin fibrosis. Identification and isolation of a dermal lineage Tassava, R.A., Nace, J.D., Wei, Y., 1996. Extracellular matrix protein turnover during
with intrinsic fibrogenic potential. Science 348, aaa2151. salamander limb regeneration. Wound Repair Regen. 4, 75–81.
Said, S., Parke, W., Neufeld, D.A., 2004. Vascular supplies differ in regenerating and Thornton, C.S., 1957a. The effect of apical cap removal on limb regeneration in
nonregenerating amputated rodent digits. Anat. Rec. A Discov. Mol. Cell Evol. Biol. Amblystoma larvae. J. Exp. Zool. Part A: Ecol. Genet. Physiol. 134, 357–381.
278, 443–449. Thornton, C.S., 1957b. The effect of apical cap removal on limb regeneration in
Sammarco, M.C., Simkin, J., Cammack, A.J., Fassler, D., Gossmann, A., Marrero, L., Amblystoma larvae. J. Exp. Zool. 134, 357–381.
Lacey, M., Van Meter, K., Muneoka, K., 2015. Hyperbaric oxygen promotes proximal Thornton, C.S., 1960. Influence of an eccentric epidermal cap on limb regeneration in
bone regeneration and organized collagen composition during digit regeneration. Amblystoma larvae. Dev. Biol. 2, 551–569.
PLoS One 10, e0140156. Thornton, C.S., Steen, T.P., 1962. Eccentric blastema formation in aneurogenic limbs of
Sammarco, M.C., Simkin, J., Fassler, D., Cammack, A.J., Wilson, A., Van Meter, K., Ambystoma larvae following epidermal cap deviation. Dev. Biol. 5, 328–343.
Muneoka, K., 2014. Endogenous bone regeneration Is dependent Upon a dynamic Thornton, C.S., Thornton, M.T., 1965. The regeneration of accessory limb parts following
oxygen event. J. Bone Miner. Res.. epidermal cap transplantation in urodeles. Experientia 21, 146–148.
Satoh, A., Graham, G.M., Bryant, S.V., Gardiner, D.M., 2008. Neurotrophic regulation of Tomlinson, B.L., Barger, P.M., 1987. A test of the punctuated-cycling hypothesis in
epidermal dedifferentiation during wound healing and limb regeneration in the Ambystoma forelimb regenerates: the roles of animal size, limb innervation, and the
axolotl (Ambystoma mexicanum). Dev. Biol. 319, 321–335. aneurogenic condition. Differentiation 35, 6–15.
Satoh, A., Hirata, A., Makanae, A., 2012. Collagen reconstitution is inversely correlated Tu, S., Johnson, S.L., 2011. Fate restriction in the growing and regenerating zebrafish fin.
with induction of limb regeneration in Ambystoma mexicanum. Zool. Sci. 29, Dev. Cell 20, 725–732.
191–197. Vinarsky, V., Atkinson, D.L., Stevenson, T.J., Keating, M.T., Odelberg, S.J., 2005. Normal
Seifert, A.W., Kiama, S.G., Seifert, M.G., Goheen, J.R., Palmer, T.M., Maden, M., 2012. newt limb regeneration requires matrix metalloproteinase function. Dev. Biol. 279,
Skin shedding and tissue regeneration in African spiny mice (Acomys). Nature 489, 86–98.
561–565. Vorontsova, M.A., Liosner, L.D., 1960. Asexual Propagation and Regeneration.
Seifert, A.W., Maden, M., 2014. New insights into vertebrate skin regeneration. Int. Rev. Pergamon Press, New York.
Cell Mol. Biol. 310, 129–169. Wallace, H., 1981. Vertebrate Limb Regeneration. John Wiley and Sons.
Simkin, J., Gawriluk, T.R., Gensel, J.C., Seifert, A.W., 2017. Macrophages are necessary Wietecha, M.S., Krol, M.J., Michalczyk, E.R., Chen, L., Gettins, P.G., DiPietro, L.A., 2015.
for epimorphic regeneration in African spiny mice. elife 6, e24623. Pigment epithelium-derived factor as a multifunctional regulator of wound healing.
Simkin, J., Sammarco, M.C., Dawson, L.A., Schanes, P.P., Yu, L., Muneoka, K., 2015a. Am. J. Physiol. Heart Circ. Physiol. 309, H812–H826.
The mammalian blastema: regeneration at our fingertips. Regeneration 2, 13. Williams-Boyce, P.K., Daniel, J.C., Jr., 1980. Regeneration of rabbit ear tissue. J. Exp.
Simkin, J., Sammarco, M.C., Dawson, L.A., Tucker, C., Taylor, L.J., Van Meter, K., Zool. 212, 243–253.
Muneoka, K., 2015b. Epidermal closure regulates histolysis during mammalian Williams-Boyce, P.K., Daniel, J.C., Jr., 1986. Comparison of ear tissue regeneration in
(Mus) digit regeneration. Regeneration 2, 14. mammals. J. Anat. 149, 55–63.
Singer, M., 1952. The influence of the nerve in regeneration of the amphibian extremity. Wu, C., Rankin, E.B., Castellini, L., Alcudia, J.F., LaGory, E.L., Andersen, R., Rhodes,
Q. Rev. Biol. 27, 169–200. S.D., Wilson, T.L., Mohammad, K.S., Castillo, A.B., Guise, T.A., Schipani, E., Giaccia,
Singh, B.N., Doyle, M.J., Weaver, C.V., Koyano-Nakagawa, N., Garry, D.J., 2012. A.J., 2015. Oxygen-sensing PHDs regulate bone homeostasis through the
Hedgehog and Wnt coordinate signaling in myogenic progenitors and regulate limb modulation of osteoprotegerin. Genes Dev. 29, 817–831.
regeneration. Dev. Biol. 371, 23–34. Yu, L., Han, M., Yan, M., Lee, E.C., Lee, J., Muneoka, K., 2010. BMP signaling induces
Stoick-Cooper, C.L., Moon, R.T., Weidinger, G., 2007. Advances in signaling in vertebrate digit regeneration in neonatal mice. Development 137, 551–559.
regeneration as a prelude to regenerative medicine. Genes Dev. 21, 1292–1315. Yu, L., Han, M., Yan, M., Lee, J., Muneoka, K., 2012. BMP2 induces segment-specific
Sugiura, T., Wang, H., Barsacchi, R., Simon, A., Tanaka, E.M., 2016. MARCKS-like skeletal regeneration from digit and limb amputations by establishing a new
protein is an initiating molecule in axolotl appendage regeneration. Nature. endochondral ossification center. Dev. Biol. 372, 263–273.
Takeo, M., Chou, W.C., Sun, Q., Lee, W., Rabbani, P., Loomis, C., Taketo, M.M., Ito, M., Yu, L., Yan, M., Simkin, J., Ketcham, P.D., Leininger, E., Han, M., Muneoka, K., 2014.
2013. Wnt activation in nail epithelium couples nail growth to digit regeneration. Angiogenesis is inhibitory for mammalian digitregeneration. Regeneration 1, 33–46.
Nature 499, 228–232.

199

View publication stats

You might also like